Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Am Chem Soc ; 146(12): 8216-8227, 2024 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-38486429

RESUMO

Bioorthogonal reactions provide a powerful tool to manipulate biological processes in their native environment. However, the transition-metal catalysts (TMCs) for bioorthogonal catalysis are limited to low atomic utilization and moderate catalytic efficiency, resulting in unsatisfactory performance in a complex physiological environment. Herein, sulfur-doped Fe single-atom catalysts with atomically dispersed and uniform active sites are fabricated to serve as potent bioorthogonal catalysts (denoted as Fe-SA), which provide a powerful tool for in situ manipulation of cellular biological processes. As a proof of concept, the N6-methyladensoine (m6A) methylation in macrophages is selectively regulated by the mannose-modified Fe-SA nanocatalysts (denoted as Fe-SA@Man NCs) for potent cancer immunotherapy. Particularly, the agonist prodrug of m6A writer METTL3/14 complex protein (pro-MPCH) can be activated in situ by tumor-associated macrophage (TAM)-targeting Fe-SA@Man, which can upregulate METTL3/14 complex protein expression and then reprogram TAMs for tumor killing by hypermethylation of m6A modification. Additionally, we find the NCs exhibit an oxidase (OXD)-like activity that further boosts the upregulation of m6A methylation and the polarization of macrophages via producing reactive oxygen species (ROS). Ultimately, the reprogrammed M1 macrophages can elicit immune responses and inhibit tumor proliferation. Our study not only sheds light on the design of single-atom catalysts for potent bioorthogonal catalysis but also provides new insights into the spatiotemporal modulation of m6A RNA methylation for the treatment of various diseases.


Assuntos
Adenosina/análogos & derivados , Imunoterapia , Neoplasias , Humanos , Metilação de RNA , Catálise , Metiltransferases
2.
J Am Chem Soc ; 145(48): 26296-26307, 2023 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-37987621

RESUMO

Immunotherapy of triple-negative breast cancer (TNBC) has an unsatisfactory therapeutic outcome due to an immunologically "cold" microenvironment. Fusobacterium nucleatum (F. nucleatum) was found to be colonized in triple-negative breast tumors and was responsible for the immunosuppressive tumor microenvironment and tumor metastasis. Herein, we constructed a bacteria-derived outer membrane vesicle (OMV)-coated nanoplatform that precisely targeted tumor tissues for dual killing of F. nucleatum and cancer cells, thus transforming intratumor bacteria into immunopotentiators in immunotherapy of TNBC. The as-prepared nanoparticles efficiently induced immunogenic cell death through a Fenton-like reaction, resulting in enhanced immunogenicity. Meanwhile, intratumoral F. nucleatum was killed by metronidazole, resulting in the release of pathogen-associated molecular patterns (PAMPs). PAMPs cooperated with OMVs further facilitated the maturation of dendritic cells and subsequent T-cell infiltration. As a result, the "kill two birds with one stone" strategy warmed up the cold tumor environment, maximized the antitumor immune response, and achieved efficient therapy of TNBC as well as metastasis prevention. Overall, this strategy based on a microecology distinction in tumor and normal tissue as well as microbiome-induced reversal of cold tumors provides new insight into the precise and efficient immune therapy of TNBC.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/metabolismo , Adjuvantes Imunológicos , Moléculas com Motivos Associados a Patógenos/metabolismo , Moléculas com Motivos Associados a Patógenos/uso terapêutico , Imunoterapia/métodos , Fusobacterium nucleatum/metabolismo , Linhagem Celular Tumoral , Microambiente Tumoral
3.
Small ; 19(37): e2207510, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37231552

RESUMO

Intracellular bacterial pathogens hiding in host cells tolerate the innate immune system and high-dose antibiotics, resulting in recurrent infections that are difficult to treat. Herein, a homing missile-like nanotherapeutic (FeSAs@Sa.M) composed of a single-atom iron nanozyme (FeSAs) core coated with infected macrophage membrane (Sa.M) is developed for in situ elimination of intracellular methicillin-resistant S. aureus (MRSA). Mechanically, the FeSAs@Sa.M initially binds to the extracellular MRSA via the bacterial recognition ability of the Sa.M component. Subsequently, the FeSAs@Sa.M can be transported to the intracellular MRSA-located regions in the host cell like a homing missile under the guidance of the extracellular MRSA to which it is attached, generating highly toxic reactive oxygen species (ROS) for intracellular MRSA killing via the enzymatic activities of the FeSAs core. The FeSAs@Sa.M is far superior to FeSAs in killing intracellular MRSA, proposing a feasible strategy for treating intracellular infections by in situ generating ROS in bacterial residing regions.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Humanos , Espécies Reativas de Oxigênio , Domínio Catalítico , Infecções Estafilocócicas/tratamento farmacológico , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico
4.
Small ; 19(32): e2301096, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37066737

RESUMO

The proper functioning of host defense system (HDS) is the key to combating bacterial infection in biological organisms. However, the delicate HDS may be dysfunctional or dysregulated, resulting in persistent infection, tissue damage, or delayed wound healing. Herein, a powerful artificial "host defense system" (aHDS) is designed and constructed for treatment of bacterial infections. First, the aHDS can quickly trap the bacteria by electrostatic interactions. Next, the system can be stimulated to produce large amounts of cytotoxic reactive oxygen species (ROS) and exert strong antibacterial effects, which can further regulate the immune microenvironment, leading to macrophage polarization from M0 to pro-inflammatory phenotype (M1) for synergistic bacteria killing. At the later stages, the system can exhibit excellent antioxidant enzyme-like activities to reprogram the M1 macrophage to anti-inflammatory phenotype (M2) for accelerating wound healing. This powerful aHDS can effectively combat the bacteria and avoid excessive inflammatory responses for the treatment of bacteria-infected wounds.


Assuntos
Infecções Bacterianas , Cicatrização , Humanos , Fenótipo , Bactérias , Antibacterianos/farmacologia , Infecções Bacterianas/tratamento farmacológico
5.
J Transl Med ; 21(1): 147, 2023 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-36829235

RESUMO

BACKGROUND: Valtrate, a natural compound isolated from the root of Valeriana, exhibits antitumor activity in many cancers through different mechanisms. However, its efficacy for the treatment of glioblastoma (GBM), a tumor type with a poor prognosis, has not yet been rigorously investigated. METHODS: GBM cell lines were treated with valtrate and CCK-8, colony formation and EdU assays, flow cytometry, and transwell, 3D tumor spheroid invasion and GBM-brain organoid co-culture invasion assays were performed to assess properties of proliferation, viability, apoptosis and invasion/migration. RNA sequencing analysis on valtrate-treated cells was performed to identify putative target genes underlying the antitumor activity of the drug in GBM cells. Western blot analysis, immunofluorescence and immunohistochemistry were performed to evaluate protein levels in valtrate-treated cell lines and in samples obtained from orthotopic xenografts. A specific activator of extracellular signal-regulated kinase (ERK) was used to identify the pathways mediating the effect. RESULTS: Valtrate significantly inhibited the proliferation of GBM cells in vitro by inducing mitochondrial apoptosis and suppressed invasion and migration of GBM cells by inhibiting levels of proteins associated with epithelial mesenchymal transition (EMT). RNA sequencing analysis of valtrate-treated GBM cells revealed platelet-derived growth factor receptor A (PDGFRA) as a potential target downregulated by the drug. Analysis of PDGFRA protein and downstream mediators demonstrated that valtrate inhibited PDGFRA/MEK/ERK signaling. Finally, treatment of tumor-bearing nude mice with valtrate led to decreased tumor volume (fivefold difference at day 28) and enhanced survival (day 27 vs day 36, control vs valtrate-treated) relative to controls. CONCLUSIONS: Taken together, our study demonstrated that the natural product valtrate elicits antitumor activity in GBM cells through targeting PDGFRA and thus provides a candidate therapeutic compound for the treatment of GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Valeriana , Camundongos , Animais , Humanos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Valeriana/metabolismo , Camundongos Nus , Proliferação de Células , Glioblastoma/patologia , Transdução de Sinais , Iridoides/farmacologia , Iridoides/uso terapêutico , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno/uso terapêutico , Linhagem Celular Tumoral , Neoplasias Encefálicas/genética , Movimento Celular
6.
Bioorg Chem ; 141: 106863, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37722269

RESUMO

Co-culturing the marine-derived fungi Penicillium janthinellium with Paecilomyces formosus led to the isolation of nine new indole-diterpenes, janthinellumines A-I (1-9), along with twelve known analogues (10-21). The chemical structures including their absolute configurations of them were assigned by the analysis of extensive spectroscopic data and calculated ECD and VCD methods. These indole-diterpenoids displayed extensive biological activities, including anti-influenza A virus, protein tyrosine phosphatase (PTP) inhibitory, and anti-Vibrio activities. Among them, the anti-influenza mechanism of compounds 1, 2, and 7 was further investigated using neuraminidase inhibitory assay, molecular docking, and reverse genetics methods, suggesting that 1, 2, and 7 could interact with Arg371 of the viral neuraminidase. The structure-activity relationship (SAR) of PTPs inhibitory activity for indole-diterpene derivatives (1, 2, 4, 5, 9-16, and 19-21) was also summarized.


Assuntos
Diterpenos , Paecilomyces , Penicillium , Simulação de Acoplamento Molecular , Técnicas de Cocultura , Neuraminidase/metabolismo , Indóis/química , Penicillium/química , Paecilomyces/metabolismo , Diterpenos/química , Estrutura Molecular
7.
Angew Chem Int Ed Engl ; 62(32): e202303989, 2023 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-37293817

RESUMO

Since polyoxometalates (POMs) can undergo reversible multi-electron redox transformations, they have been used to modulate the electronic environment of metal nanoparticles for catalysis. Besides, POMs possess unique electronic structures and acid-responsive self-assembly ability. These properties inspired us to tackle the drawbacks of the copper-catalyzed azide-alkyne cycloaddition (CuAAC) reaction in biomedical applications, such as low catalytic efficiency and unsatisfactory disease selectivity. Herein, we construct molybdenum (Mo)-based POM nanoclusters doped with Cu (Cu-POM NCs) as a highly efficient bioorthogonal catalyst, which is responsive to pathologicallyacid and H2 S for selective antibiofilm therapy. Leveraging the merits of POMs, the Cu-POM NCs exhibit biofilm-responsive self-assembly behavior, efficient CuAAC-mediated in situ synthesis of antibacterial molecules, and a NIR-II photothermal effect selectively triggered by H2 S in pathogens. The consumption of bacterial H2 S at the pathological site by Cu-POM NCs extremely decreases the number of persisterbacteria, which is conducive to the inhibition of bacterial tolerance and elimination of biofilms. Unlocked at pathological sites and endowed with NIR-II photothermal property, the constructed POM-based bioorthogonal catalytic platform provides new insights into the design of efficient and selective bioorthogonal catalysts for disease therapy.


Assuntos
Cobre , Molibdênio , Cobre/química , Molibdênio/química , Catálise , Alcinos/química
8.
J Pharmacol Exp Ther ; 380(1): 63-75, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34750208

RESUMO

Thiabendazole (TBZ), approved by the US Food and Drug Administration (FDA) for human oral use, elicits a potential anticancer activity on cancer cells in vitro and in animal models. Here, we evaluated the efficacy of TBZ in the treatment of human glioblastoma multiforme (GBM). TBZ reduced the viability of GBM cells (P3, U251, LN229, A172, and U118MG) relative to controls in a dose- and time-dependent manner. However, normal human astrocytes (NHA) exhibited a greater IC50 than tumor cell lines and were thus more resistant to its cytotoxic effects. 5-Ethynyl-2'-deoxyuridine (EdU)-positive cells and the number of colonies formed were decreased in TBZ-treated cells (at 150 µM, P < 0.05 and at 150 µM, P < 0.001, respectively). This decrease in proliferation was associated with a G2/M arrest as assessed with flow cytometry, and the downregulation of G2/M check point proteins. In addition, TBZ suppressed GBM cell invasion. Analysis of RNA sequencing data comparing TBZ-treated cells with controls yielded a group of differentially expressed genes, the functions of which were associated with the cell cycle and DNA replication. The most significantly downregulated gene in TBZ-treated cells was mini-chromosome maintenance protein 2 (MCM2). SiRNA knockdown of MCM2 inhibited proliferation, causing a G2/M arrest in GBM cell lines and suppressed invasion. Taken together, our results demonstrated that TBZ inhibited proliferation and invasion in GBM cells through targeting of MCM2. SIGNIFICANCE STATEMENT: TBZ inhibits the proliferation and invasion of glioblastoma cells by downregulating the expression of MCM2. These results support the repurposing of TBZ as a possible therapeutic drug in the treatment of GBM.


Assuntos
Anti-Helmínticos/uso terapêutico , Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Componente 2 do Complexo de Manutenção de Minicromossomo/metabolismo , Tiabendazol/farmacologia , Animais , Anti-Helmínticos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Reposicionamento de Medicamentos , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos Nus , Tiabendazol/uso terapêutico
9.
J Stroke Cerebrovasc Dis ; 29(3): 104601, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31917091

RESUMO

Herein, we described 2 patients with posterior spinal artery syndrome (PSAS) caused by vertebral artery dissection. The patients complained of sudden neck pain or walking instability. Neurological examination revealed sensory loss, muscle weakness, and sensory ataxia. Angiography showed double lumen sign or intimal flap in the vertebral artery. T2-weighted imaging and diffusion-weighted imaging of MRI showed a hyperintense lesion in the dorsal side of the cervical spinal cord at different times after onset. Both patients had good outcome after antiplatelet therapy and physiotherapy. A review of previously reported PSAS cases was also conducted in order to improve the understanding and awareness of this rare myelopathy.


Assuntos
Doenças Vasculares da Medula Espinal/etiologia , Dissecação da Artéria Vertebral/complicações , Adulto , Feminino , Marcha , Transtornos Neurológicos da Marcha/etiologia , Transtornos Neurológicos da Marcha/fisiopatologia , Transtornos Neurológicos da Marcha/terapia , Humanos , Masculino , Cervicalgia/etiologia , Cervicalgia/fisiopatologia , Cervicalgia/terapia , Modalidades de Fisioterapia , Inibidores da Agregação Plaquetária/uso terapêutico , Recuperação de Função Fisiológica , Doenças Vasculares da Medula Espinal/diagnóstico por imagem , Doenças Vasculares da Medula Espinal/fisiopatologia , Doenças Vasculares da Medula Espinal/terapia , Resultado do Tratamento , Dissecação da Artéria Vertebral/diagnóstico por imagem , Dissecação da Artéria Vertebral/fisiopatologia , Dissecação da Artéria Vertebral/terapia
10.
J Virol ; 90(4): 1910-7, 2016 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-26656689

RESUMO

UNLABELLED: Coronaviruses (CoVs) can cause highly prevalent diseases in humans and animals. Feline infectious peritonitis virus (FIPV) belongs to the genus Alphacoronavirus, resulting in a lethal systemic granulomatous disease called feline infectious peritonitis (FIP), which is one of the most important fatal infectious diseases of cats worldwide. No specific vaccines or drugs have been approved to treat FIP. CoV main proteases (M(pro)s) play a pivotal role in viral transcription and replication, making them an ideal target for drug development. Here, we report the crystal structure of FIPV M(pro) in complex with dual inhibitors, a zinc ion and a Michael acceptor. The complex structure elaborates a unique mechanism of two distinct inhibitors synergizing to inactivate the protease, providing a structural basis to design novel antivirals and suggesting the potential to take advantage of zinc as an adjunct therapy against CoV-associated diseases. IMPORTANCE: Coronaviruses (CoVs) have the largest genome size among all RNA viruses. CoV infection causes various diseases in humans and animals, including severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS). No approved specific drugs or vaccinations are available to treat their infections. Here, we report a novel dual inhibition mechanism targeting CoV main protease (M(pro)) from feline infectious peritonitis virus (FIPV), which leads to lethal systemic granulomatous disease in cats. M(pro), conserved across all CoV genomes, is essential for viral replication and transcription. We demonstrated that zinc ion and a Michael acceptor-based peptidomimetic inhibitor synergistically inactivate FIPV M(pro). We also solved the structure of FIPV M(pro) complexed with two inhibitors, delineating the structural view of a dual inhibition mechanism. Our study provides new insight into the pharmaceutical strategy against CoV M(pro) through using zinc as an adjuvant therapy to enhance the efficacy of an irreversible peptidomimetic inhibitor.


Assuntos
Coronavirus Felino/enzimologia , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Inibidores de Proteases/química , Inibidores de Proteases/metabolismo , Sequência de Aminoácidos , Proteases 3C de Coronavírus , Cristalografia por Raios X , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Zinco/química , Zinco/metabolismo
11.
CNS Neurosci Ther ; 30(2): e14366, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37485655

RESUMO

AIMS: PSMD family members, as important components of the 26S proteasome, are well known to be involved in protein degradation. However, their role in glioblastoma (GBM) has not been rigorously investigated. We aimed to perform systematic analysis of the expression signature, prognostic significance and functions of PSMD family genes in GBM to reveal potential prognostic markers and new therapeutic targets among PSMD family members. METHODS: In this study, we systemically analyzed PSMD family members in terms of their expression profiles, prognostic implications, DNA methylation levels, and genetic alterations; the relationships between their expression levels and immune infiltration and drug sensitivity; and their potential functional enrichment in GBM through bioinformatics assessment. Moreover, in vitro and in vivo experiments were used to validate the biological functions of PSMD9 and its targeted therapeutic effect in GBM. RESULTS: The mRNA levels of PSMD5/8/9/10/11/13/14 were higher in GBM than in normal brain tissues, and the mRNA levels of PSMD1/4/5/8/9/11/12 were higher in high-grade glioma (WHO grade III & IV) than in low-grade glioma (WHO grade II). High mRNA expression of PSMD2/6/8/9/12/13/14 and low mRNA expression of PSMD7 were associated with poor overall survival (OS). Multivariate Cox regression analysis identified PSMD2/5/6/8/9/10/11/12 as independent prognostic factors for OS prediction. In addition, the protein-protein interaction network and gene set enrichment analysis results suggested that PSMD family members and their interacting molecules were involved in the regulation of the cell cycle, cell invasion and migration, and other biological processes in GBM. In addition, knockdown of PSMD9 inhibited cell proliferation, invasion and migration and induced G2/M cell cycle arrest in LN229 and A172 GBM cells. Moreover, PSMD9 promoted the malignant progression of GBM in vivo. GBM cell lines with high PSMD9 expression were more resistant to panobinostat, a potent deacetylase inhibitor, than those with low PSMD9 expression. In vitro and in vivo experiments further validated that PSMD9 overexpression rescued the GBM inhibitory effect of panobinostat. CONCLUSION: This study provides new insights into the value of the PSMD family in human GBM diagnosis and prognosis evaluation, and we further identified PSMD9 as a potential therapeutic target. These findings may lead to the development of effective therapeutic strategies for GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Panobinostat , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Glioma/genética , Prognóstico , Fatores de Transcrição/genética , RNA Mensageiro/metabolismo , Regulação Neoplásica da Expressão Gênica , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo
12.
Pest Manag Sci ; 2024 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-39109536

RESUMO

BACKGROUND: The pine wood nematode Bursaphelenchus xylophilus, a severe invasive species, is responsible for causing widespread pine wilt disease. The CytCo protein, a pore-forming toxin derived from Conidiobolus obscurus, exhibits nematotoxicity towards B. xylophilus. RESULTS: Our present study reveals the expression variation of a range of gene products in B. xylophilus that respond to the effects of CytCo using the isobaric tags for relative and absolute quantification proteomics technology. Functional enrichment analysis indicates that many differentially expressed proteins are linked to calcium signaling system, proteasome, energy production and conversion, and the determination of adult lifespan. It suggests that the dysregulation of calcium homeostasis, energy metabolism, and apoptosis contribute to the CytCo nematotoxicity. Using the calcium ion (Ca2+)-indicator calcein, we detected changes in Ca2+ levels in B. xylophilus, with a significantly increase in fluorescence in the nematode's intestine and pseudocoelom following CytCo treatments. Meanwhile, the apoptosis and reactive oxygen species (ROS) assays showed an enhancement of fluorescence in B. xylophilus cells, with increased CytCo concentrations. CONCLUSION: The protein toxin CytCo triggers Ca2+ leakage, disrupts Ca2+ balance in B. xylophilus, and induces apoptosis and ROS outburst, thereby intensifying its nematotoxic effects. This finding facilitates our understanding of the modes of action of nematotoxic proteins, and contributes to the development of innovative nematode control strategies. © 2024 Society of Chemical Industry.

13.
Nat Prod Bioprospect ; 14(1): 48, 2024 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-39158647

RESUMO

Recently the FDA conducted a risk investigation and labeled the Boxed Warning for all BCMA- and CD19-directed CAR-T cell therapy, so does it mean that the public must take risk of secondary cancer to receive cell therapy? Here, without lentivirus and professional antigen presenting cell application, a novel tumor-specific T-cell therapy was successfully developed only by co-culturing MHC+ cancer cells and Naïve-T cells under the CD28 co-stimulatory signals. These tumor-specific T-cells could be separated through cell size and abundantly produced from peripheral blood, and would spontaneously attack target cells that carrying the same tumor antigen while avoiding others in vitro test. Moreover, it markedly decreased 90% tumor nodules companying with greatly improving overall survival (76 days vs 30 days) after twice infusion back to mice. This work maximally avoided the risks of secondary cancer and non-specific killing, and might open a revolutionary beginning of natural tumor-specific T-cell therapy.

14.
Adv Mater ; 36(14): e2310063, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38153294

RESUMO

Pyroptosis has garnered increasing attention in cancer immunotherapy. Moreover, increasing plasma membrane damage by reactive oxygen species (ROS) is considered an effective strategy for promoting pyroptosis. However, the current tactics for enhancing membrane rupture in pyroptosis are limited by the inherent drawbacks of ROS and the immunosuppressive tumor microenvironment. Herein, a self-adaptive pyroptosis inducer (LPZ) is designed by integrating Lactobacillus rhamnosus GG (LGG) and an enzyme-like metal-organic framework to achieve potent pyroptosis immunotherapy. LPZ can adhere to cancer cell membranes through the interaction between the pili of LGG and the mucin of cancer cells. In particular, the adaptive formula can gradually enhance the ability of nanozymes to produce ROS by creating an acidic microenvironment through anaerobic respiration. These results verify that LPZ could generate high levels of ROS both on the membrane and within cancer cells, leading to pyroptotic cell death and strong antitumor immunity. Meanwhile, LGG are eventually killed by ROS in this process to halt their respiration and prevent potential biosafety concerns. Overall, this work provides new inspiration for the design of self-adaptive nanocatalytic drugs for cancer immunotherapy.


Assuntos
Neoplasias , Piroptose , Humanos , Espécies Reativas de Oxigênio , Membrana Celular , Catálise , Imunoterapia , Microambiente Tumoral , Neoplasias/terapia
15.
Int J Biol Macromol ; 279(Pt 3): 135401, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39245109

RESUMO

The pine wood nematode Bursaphelenchus xylophilus is a highly invasive species responsible for the widespread pine wilt disease. Double-stranded RNA (dsRNA) biopesticides represent a novel strategy for controlling plant-parasitic nematodes. The B. xylophilus arginine kinase (BxAK) features a conserved ATP-binding domain and exhibits nematode-specific divergence in the phylogenetic tree. Notably, whole-mount in situ hybridization signals are evident in the nematode head and middle sections, particularly in the juvenile stage before sex differentiation. In this study, we developed a novel dsRNA-like small interfering RNA (siRNA) assembly that specifically targets BxAK and presents highly nematicidal effects. The RNA interference (RNAi) efficiency achieved a 95.9 % reduction in second-stage juveniles. In bioassays, the median lethal concentrations of this siRNA assembly against B. xylophilus were 168.5 ng/µl for juveniles and 603.8 ng/µl for adults within 48 h. Moreover, transcriptomic results revealed significantly downregulated expression levels of genes related to metabolism and development, suggesting that the mode of action of BxAK silencing is related to disruptions in energy homeostasis and juvenile development. In conclusion, BxAK is a molecular target for controlling B. xylophilus, and our siRNA assembly significantly enhances RNAi efficiency and lowers the lethal concentration required, making it a promising candidate for future biocontrol applications.


Assuntos
Arginina Quinase , Pinus , Interferência de RNA , RNA de Cadeia Dupla , RNA Interferente Pequeno , Animais , RNA de Cadeia Dupla/genética , Arginina Quinase/genética , Arginina Quinase/metabolismo , RNA Interferente Pequeno/genética , Pinus/parasitologia , Antinematódeos/farmacologia , Tylenchida/genética , Tylenchida/enzimologia , Inativação Gênica , Filogenia , Doenças das Plantas/parasitologia , Doenças das Plantas/genética , Proteínas de Helminto/genética , Proteínas de Helminto/metabolismo
16.
Neuro Oncol ; 26(4): 653-669, 2024 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-38069906

RESUMO

BACKGROUND: Altered branched-chain amino acid (BCAA) metabolism modulates epigenetic modification, such as H3K27ac in cancer, thus providing a link between metabolic reprogramming and epigenetic change, which are prominent hallmarks of glioblastoma multiforme (GBM). Here, we identified mitochondrial 3-hydroxymethyl-3-methylglutaryl-CoA lyase (HMGCL), an enzyme involved in leucine degradation, promoting GBM progression and glioma stem cell (GSC) maintenance. METHODS: In silico analysis was performed to identify specific molecules involved in multiple processes. Glioblastoma multiforme cells were infected with knockdown/overexpression lentiviral constructs of HMGCL to assess malignant performance in vitro and in an orthotopic xenograft model. RNA sequencing was used to identify potential downstream molecular targets. RESULTS: HMGCL, as a gene, increased in GBM and was associated with poor survival in patients. Knockdown of HMGCL suppressed proliferation and invasion in vitro and in vivo. Acetyl-CoA was decreased with HMGCL knockdown, which led to reduced NFAT1 nuclear accumulation and H3K27ac level. RNA sequencing-based transcriptomic profiling revealed FOXM1 as a candidate downstream target, and HMGCL-mediated H3K27ac modification in the FOXM1 promoter induced transcription of the gene. Loss of FOXM1 protein with HMGCL knockdown led to decreased nuclear translocation and thus activity of ß-catenin, a known oncogene. Finally, JIB-04, a small molecule confirmed to bind to HMGCL, suppressed GBM tumorigenesis in vitro and in vivo. CONCLUSIONS: Changes in acetyl-CoA levels induced by HMGCL altered H3K27ac modification, which triggers transcription of FOXM1 and ß-catenin nuclear translocation. Targeting HMGCL by JIB-04 inhibited tumor growth, indicating that mediators of BCAA metabolism may serve as molecular targets for effective GBM treatment.


Assuntos
Aminopiridinas , Glioblastoma , Hidrazonas , Liases , Humanos , Acetilcoenzima A/genética , Acetilcoenzima A/metabolismo , Acetilação , beta Catenina/genética , Linhagem Celular Tumoral , Proliferação de Células , Proteína Forkhead Box M1/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Histonas/genética , Liases/genética , Liases/metabolismo
17.
J Exp Clin Cancer Res ; 43(1): 25, 2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38246990

RESUMO

BACKGROUND: Extensive local invasion of glioblastoma (GBM) cells within the central nervous system (CNS) is one factor that severely limits current treatments. The aim of this study was to uncover genes involved in the invasion process, which could also serve as therapeutic targets. For the isolation of invasive GBM cells from non-invasive cells, we used a three-dimensional organotypic co-culture system where glioma stem cell (GSC) spheres were confronted with brain organoids (BOs). Using ultra-low input RNA sequencing (ui-RNA Seq), an invasive gene signature was obtained that was exploited in a therapeutic context. METHODS: GFP-labeled tumor cells were sorted from invasive and non-invasive regions within co-cultures. Ui-RNA sequencing analysis was performed to find a gene cluster up-regulated in the invasive compartment. This gene cluster was further analyzed using the Connectivity MAP (CMap) database. This led to the identification of SKF83566, an antagonist of the D1 dopamine receptor (DRD1), as a candidate therapeutic molecule. Knockdown and overexpression experiments were performed to find molecular pathways responsible for the therapeutic effects of SKF83566. Finally, the effects of SKF83566 were validated in orthotopic xenograft models in vivo. RESULTS: Ui-RNA seq analysis of three GSC cell models (P3, BG5 and BG7) yielded a set of 27 differentially expressed genes between invasive and non-invasive cells. Using CMap analysis, SKF83566 was identified as a selective inhibitor targeting both DRD1 and DRD5. In vitro studies demonstrated that SKF83566 inhibited tumor cell proliferation, GSC sphere formation, and invasion. RNA sequencing analysis of SKF83566-treated P3, BG5, BG7, and control cell populations yielded a total of 32 differentially expressed genes, that were predicted to be regulated by c-Myc. Of these, the UHRF1 gene emerged as the most downregulated gene following treatment, and ChIP experiments revealed that c-Myc binds to its promoter region. Finally, SKF83566, or stable DRD1 knockdown, inhibited the growth of orthotopic GSC (BG5) derived xenografts in nude mice. CONCLUSIONS: DRD1 contributes to GBM invasion and progression by regulating c-Myc entry into the nucleus that affects the transcription of the UHRF1 gene. SKF83566 inhibits the transmembrane protein DRD1, and as such represents a candidate small therapeutic molecule for GBMs.


Assuntos
Antagonistas de Dopamina , Glioblastoma , Glioma , Proteínas Proto-Oncogênicas c-myc , Animais , Humanos , Camundongos , Encéfalo , Proteínas Estimuladoras de Ligação a CCAAT/efeitos dos fármacos , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Dopamina , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Camundongos Nus , Família Multigênica , Receptores de Dopamina D1/antagonistas & inibidores , Ubiquitina-Proteína Ligases/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo , Antagonistas de Dopamina/metabolismo , Antagonistas de Dopamina/farmacologia , Proteínas Proto-Oncogênicas c-myc/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/metabolismo
18.
Environ Sci Pollut Res Int ; 30(6): 14484-14496, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36153423

RESUMO

Improving industrial eco-efficiency is of great significance for building a beautiful China and achieving its carbon peak and neutrality targets. Based on the panel data of 30 provinces in China from 2007 to 2018, this paper uses the super-efficiency SBM model to measure industrial eco-efficiency and empirically tests the influence of green finance on Chinese industrial eco-efficiency from the national and regional levels. The results show that the average level of industrial eco-efficiency in China is relatively stable during the study period with a large space for advancement. Second, there is spatial heterogeneity in Chinese industrial eco-efficiency, showing a gradually decreasing "southeast-northwest" ladder-like distribution. Third, the national-level regression results show that there is a significant "U-shaped" relationship between green financing and industrial eco-efficiency. In addition, the regression results at the regional level indicate that there is regional heterogeneity in the impact of green finance on industrial eco-efficiency. Finally, based on the research conclusions, specific suggestions on how green finance can improve industrial eco-efficiency in China are put forward, including vigorously developing green finance at the macro and micro levels, and exerting the positive effects of green finance in improving industrial eco-efficiency according to the area and the development level of green finance.


Assuntos
Eficiência , Indústrias , China , Carbono , Desenvolvimento Econômico
19.
Artigo em Inglês | MEDLINE | ID: mdl-37691061

RESUMO

Digital inclusive finance (DIF) provides new momentum for green agricultural development (AGD). This paper measured AGD with entropy weight TOPSIS in five dimensions, including resource conservation, environmental friendliness, ecological conservation, green supply, and economic growth. After that, it estimated the regional spillover effects and threshold impacts of DIF on AGD utilizing China's provincial panel data from 2011 to 2020. The paper shows that (1) DIF and AGD have such a U-shaped complex interrelationship; (2) the AGD is spatially impacted by DIF. The unique manifestation is that as DIF has increased, its effect on AGD has steadily changed from being direct to being indirect, and this effect has regional heterogeneity; and (3) in regions with higher levels of green technology innovation, better development of traditional finance, or relatively concentrated agricultural industries, DIF plays a more prominent role in promoting the AGD.

20.
Chem Sci ; 14(6): 1598-1605, 2023 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-36794177

RESUMO

Conventional strategies for treating inflammatory bowel disease merely relieve inflammation and excessive immune response, but fail to solve the underlying causes of IBD, such as disrupted gut microbiota and intestinal barrier. Recently, natural probiotics have shown tremendous potential for the treatment of IBD. However, probiotics are not recommended for IBD patients, as they may cause bacteremia or sepsis. Herein, for the first time, we constructed artificial probiotics (Aprobiotics) based on artificial enzyme-dispersed covalent organic frameworks (COFs) as the "organelle" and a yeast shell as the membrane of the Aprobiotics to manage IBD. The COF-based artificial probiotics, with the function of natural probiotics, could markedly relieve IBD by modulating the gut microbiota, suppressing intestinal inflammation, protecting the intestinal epithelial cells, and regulating immunity. This nature-inspired approach may aid in the design of more artificial systems for the treatment of various incurable diseases, such as multidrug-resistant bacterial infection, cancer, and others.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA